Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
J Biol Chem ; 295(34): 12188-12202, 2020 08 21.
Article in English | MEDLINE | ID: mdl-32467227

ABSTRACT

Triple-negative breast cancer (TNBC) is an aggressive cancer subtype for which effective therapies are unavailable. TNBC has a high frequency of tumor protein p53 (Tp53/p53)- and phosphatase and tensin homolog (PTEN) deficiencies, and combined p53- and PTEN-deficiency is associated with poor prognosis and poor response to anticancer therapies. In this study, we discovered that combined p53- and PTEN-deficiency in TNBC activates expression of the transcription factor mesenchyme homeobox 1 (MEOX1). We found that MEOX1 is expressed only in TNBC cells with frequent deficiencies in p53 and PTEN, and that its expression is undetectable in luminal A, luminal B, and HER2+ subtypes, as well as in normal breast cells with wild-type (WT) p53 and PTEN. Notably, siRNA knockdown of both p53 and PTEN activated MEOX1 expression in breast cancer cells, whereas individual knockdowns of either p53 or PTEN had only minimal effects on MEOX1 expression. MEOX1 knockdown abolished cell proliferation of p53- and PTEN-deficient TNBC in vitro and inhibited tumor growth in vivo, but had no effect on the proliferation of luminal and HER2+ cancer cells and normal breast cells. RNA-Seq and immunoblotting analyses showed that MEOX1 knockdown decreased expression of tyrosine kinase 2 (TYK2), signal transducer and activator of transcription 5B (STAT5B), and STAT6 in p53- and PTEN-deficient TNBC cells. These results reveal the effects of combined p53- and PTEN-deficiency on MEOX1 expression and TNBC cell proliferation, suggesting that MEOX1 may serve as a potential therapeutic target for managing p53- and PTEN-deficient TNBC.


Subject(s)
Cell Proliferation , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/biosynthesis , PTEN Phosphohydrolase/deficiency , Transcription Factors/biosynthesis , Tumor Suppressor Protein p53/deficiency , Animals , Female , Homeodomain Proteins/genetics , Humans , Mice , Mice, Inbred NOD , Mice, SCID , PTEN Phosphohydrolase/metabolism , STAT5 Transcription Factor/biosynthesis , STAT5 Transcription Factor/genetics , STAT6 Transcription Factor/biosynthesis , STAT6 Transcription Factor/genetics , TYK2 Kinase/biosynthesis , TYK2 Kinase/genetics , Transcription Factors/genetics , Triple Negative Breast Neoplasms , Tumor Suppressor Protein p53/metabolism
2.
J Biol Chem ; 293(6): 2125-2136, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29263092

ABSTRACT

The transcription factor BCL11A has recently been reported to be a driving force in triple-negative breast cancer (TNBC), contributing to the maintenance of a chemoresistant breast cancer stem cell (BCSC) population. Although BCL11A was shown to suppress γ-globin and p21 and to induce MDM2 expression in the hematopoietic system, its downstream targets in TNBC are still unclear. For its role in transcriptional repression, BCL11A was found to interact with several corepressor complexes; however, the mechanisms underlying these interactions remain unknown. Here, we reveal that BCL11A interacts with histone methyltransferase (PRC2) and histone deacetylase (NuRD and SIN3A) complexes through their common subunit, RBBP4/7. In fluorescence polarization assays, we show that BCL11A competes with histone H3 for binding to the negatively charged top face of RBBP4. To define that interaction, we solved the crystal structure of RBBP4 in complex with an N-terminal peptide of BCL11A (residues 2-16, BCL11A(2-16)). The crystal structure identifies novel interactions between BCL11A and the side of the ß-propeller of RBBP4 that are not seen with histone H3. We next show that BCL11A(2-16) pulls down RBBP4, RBBP7, and other components of PRC2, NuRD, and SIN3A from the cell lysate of the TNBC cell line SUM149. Furthermore, we demonstrate the therapeutic potential of targeting the RBBP4-BCL11A binding by showing that a BCL11A peptide can decrease aldehyde dehydrogenase-positive BCSCs and mammosphere formation capacity in SUM149. Together, our findings have uncovered a previously unidentified mechanism that BCL11A may use to recruit epigenetic complexes to regulate transcription and promote tumorigenesis.


Subject(s)
Carrier Proteins/metabolism , Nuclear Proteins/metabolism , Retinoblastoma-Binding Protein 4/metabolism , Retinoblastoma-Binding Protein 7/metabolism , Carcinogenesis , Carrier Proteins/chemistry , Cell Line , Crystallography, X-Ray , Epigenomics , Histone Deacetylases/metabolism , Histone Methyltransferases/metabolism , Humans , Nuclear Proteins/chemistry , Protein Binding , Repressor Proteins , Retinoblastoma-Binding Protein 4/chemistry , Retinoblastoma-Binding Protein 7/chemistry , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/pathology
3.
Sensors (Basel) ; 19(11)2019 Jun 10.
Article in English | MEDLINE | ID: mdl-31185624

ABSTRACT

In light of the need for Extramural Hospital Information System (HIS) access through mobile devices outside the hospital, this research analyzes situational information security threats, including the circumstances in which a mobile device may get lost and personal data may be stolen. Moreover, the system needs to be implemented in accordance with the regulations. Based on the security threat analysis, it is proposed to use a security control module to provide a security-enabled HIS proxy module, two-way authentication module, and One-Time Password (OTP). The sending module and cryptographic technology computing module with Micro SD encryption card form a set of HIS extension system, which includes the SMS OTP method to simultaneously verify the two-way authentication mechanism of a user and the device that the user owns.


Subject(s)
Cell Phone , Computer Security , Health Records, Personal , Hospital Information Systems , Algorithms , Confidentiality , Humans , Telemedicine
4.
Law Hum Behav ; 38(4): 367-77, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24707909

ABSTRACT

The first mock jury study in Taiwan, in which 279 community members watched a videotaped trial, investigated how jurors' estimates of the relative undesirability of wrongful conviction versus wrongful acquittal predicted individual decisions and how decision rules affected outcomes. The percentage of jurors who viewed wrongful conviction as more undesirable increased from 50.9% to 60.9% after deliberation and jurors' postdeliberation acquittal rate (71.7%) was higher than predeliberation acquittal rate (58.8%). Jurors' estimates of the undesirability of wrongful conviction were not correlated with their predeliberation votes but became positively correlated with their postdeliberation decisions. The unanimous rule facilitated jurors' change of vote, predominantly from conviction to acquittal, than the simple majority rule. Jurors reaching a verdict under the unanimous rule viewed deliberation and the verdict more positively. This study indicates that deliberation can ameliorate the problem of most Taiwanese citizens not viewing wrongful conviction as more undesirable than wrongful acquittal. It also suggests that Taiwan should adopt a unanimous rule for its proposed lay participation system.


Subject(s)
Criminal Law , Decision Making , Adult , Female , Humans , Male , Middle Aged , Taiwan , Young Adult
5.
Cancer Lett ; : 217219, 2024 Sep 05.
Article in English | MEDLINE | ID: mdl-39244005

ABSTRACT

Luminal Androgen Receptor (LAR) triple-negative breast cancers (TNBC) express androgen receptors (AR), exhibit high frequency of PIK3CA mutations and intact RB. Herein, we investigated combined blockade of the CDK4/6 and PI3K signaling with palbociclib, alpelisib, and capivasertib, which inhibit CDK4/6, PI3Kα, and AKT1-3, respectively. The combination of palbociclib/capivasertib, but not palbociclib/alpelisib, synergistically inhibited proliferation of MDA-MB-453 and MFM-223 LAR cells [synergy score 7.34 (p=5.81x10-11) and 4.78 (p=0.012), respectively]. The AR antagonist enzalutamide was inactive against MDA-MB-453, MFM-223, and CAL148 cells and did not enhance the efficacy of either combination. Palbociclib/capivasertib inhibited growth of LAR patient-derived xenografts more potently than palbociclib/alpelisib. Treatment of LAR cells with palbociclib suppressed phosphorylated-RB and resulted in adaptive phosphorylation/activation of S473 pAKT and AKT substrates GSK3ß, PRAS40, and FoxO3a. Capivasertib blocked palbociclib-induced phosphorylation of AKT substrates more potently than alpelisib. Treatment with PI3Kß inhibitors did not block phosphorylation of AKT substrates, suggesting that PI3Kß did not mediate the adaptive response to CDK4/6 inhibition. Phosphokinase arrays of MDA-MB-453 cells treated with palbociclib showed time-dependent upregulation of PDGFRß, GSK3ß, STAT3, and STAT6. RNA silencing of PDGFRß in palbociclib-treated MDA-MB-453 and MFM-223 cells blocked the upregulation of S473 pAKT, suggesting that the adaptive response to CDK4/6 blockade involves PDGFRß signaling. Finally, treatment with palbociclib and the PDGFR inhibitor CP637451 arrested growth of MDA-MB-453 and MFM-223 cells to the same degree as palbociclib/capivasertib. These findings support testing the combination of CDK4/6 and AKT inhibitors in patients with LAR TNBC, and further investigation of PDGFR antagonists in this breast cancer subtype.

6.
Nat Commun ; 15(1): 2287, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38480701

ABSTRACT

CDK4/6 inhibitors (CDK4/6i) have improved survival of patients with estrogen receptor-positive (ER+) breast cancer. However, patients treated with CDK4/6i eventually develop drug resistance and progress. RB1 loss-of-function alterations confer resistance to CDK4/6i, but the optimal therapy for these patients is unclear. Through a genome-wide CRISPR screen, we identify protein arginine methyltransferase 5 (PRMT5) as a molecular vulnerability in ER+/RB1-knockout breast cancer cells. Inhibition of PRMT5 blocks the G1-to-S transition in the cell cycle independent of RB, leading to growth arrest in RB1-knockout cells. Proteomics analysis uncovers fused in sarcoma (FUS) as a downstream effector of PRMT5. Inhibition of PRMT5 results in dissociation of FUS from RNA polymerase II, leading to hyperphosphorylation of serine 2 in RNA polymerase II, intron retention, and subsequent downregulation of proteins involved in DNA synthesis. Furthermore, treatment with the PRMT5 inhibitor pemrametostat and a selective ER degrader fulvestrant synergistically inhibits growth of ER+/RB-deficient cell-derived and patient-derived xenografts. These findings highlight dual ER and PRMT5 blockade as a potential therapeutic strategy to overcome resistance to CDK4/6i in ER+/RB-deficient breast cancer.


Subject(s)
Breast Neoplasms , Humans , Female , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , RNA Polymerase II , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase Inhibitor Proteins , Cyclin-Dependent Kinase 6/genetics , Cyclin-Dependent Kinase 6/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Drug Resistance, Neoplasm/genetics , Protein-Arginine N-Methyltransferases/genetics , Protein-Arginine N-Methyltransferases/metabolism
7.
Res Sq ; 2023 Jul 10.
Article in English | MEDLINE | ID: mdl-37502925

ABSTRACT

CDK4/6 inhibitors (CDK4/6i) have improved survival of patients with estrogen receptor-positive (ER+) breast cancer. However, patients treated with CDK4/6i eventually develop drug resistance and progress. RB1 loss-of-function alterations confer acquired resistance to CDK4/6i, but the optimal therapy for these patients is unclear. Using a genome-wide CRISPR screen, we identified protein arginine methyltransferase 5 (PRMT5) as a molecular vulnerability in ER+/RB1-knockout (RBKO) breast cancer cells. PRMT5 inhibition blocked cell cycle G1-to-S transition independent of RB, thus arresting growth of RBKO cells. Proteomics analysis uncovered fused in sarcoma (FUS) as a downstream effector of PRMT5. Pharmacological inhibition of PRMT5 resulted in dissociation of FUS from RNA polymerase II (Pol II), Ser2 Pol II hyperphosphorylation, and intron retention in genes that promote DNA synthesis. Treatment with the PRMT5i inhibitor pemrametostat and fulvestrant synergistically inhibited growth of ER+/RB-deficient patient-derived xenografts, suggesting dual ER and PRMT5 blockade as a novel therapeutic strategy to treat ER+/RB-deficient breast cancer.

8.
Article in English | MEDLINE | ID: mdl-22988478

ABSTRACT

The consumption of Chinese herbal medicines (CHMs) is increasing exponentially. Many patients utilize CHMs concomitantly with prescription drugs in great frequency. Herb-drug interaction has hence become an important focus of study. Transporter-mediated herb-drug interactions have the potential to seriously influence drug efficacy and toxicity. Since organic anion transporter 1 (OAT1) is crucial in renal active secretion and drug-drug interactions, the possibility of modulation of OAT1-mediated drug transport should be seriously concerned. Sixty-three clinically used CHMs were evaluated in the study. An hOAT1-overexpressing cell line was used for the in vitro CHMs screening, and the effective candidates were administered to Wistar rats to access renal hemodynamics. The regulation of OAT1 mRNA expression was also examined for further evidence of CHMs affecting OAT1-mediated transport. Among all the 63 CHMs, formulae Gui Zhi Fu Ling Wan (GZ) and Chia Wei Hsiao Yao San (CW) exhibited significant inhibitions on hOAT1-mediated [(3)H]-PAH uptake in vitro and PAH clearance and net secretion in vivo. Moreover, GZ showed concentration-dependent manners both in vitro and in vivo, and the decrease of rOAT1 mRNA expression indicated that GZ not only inhibited function of OAT1 but also suppressed expression of OAT1.

9.
Cancer Immunol Res ; 10(7): 829-843, 2022 07 01.
Article in English | MEDLINE | ID: mdl-35561311

ABSTRACT

The MYC oncogene is frequently amplified in triple-negative breast cancer (TNBC). Here, we show that MYC suppression induces immune-related hallmark gene set expression and tumor-infiltrating T cells in MYC-hyperactivated TNBCs. Mechanistically, MYC repressed stimulator of interferon genes (STING) expression via direct binding to the STING1 enhancer region, resulting in downregulation of the T-cell chemokines CCL5, CXCL10, and CXCL11. In primary and metastatic TNBC cohorts, tumors with high MYC expression or activity exhibited low STING expression. Using a CRISPR-mediated enhancer perturbation approach, we demonstrated that MYC-driven immune evasion is mediated by STING repression. STING repression induced resistance to PD-L1 blockade in mouse models of TNBC. Finally, a small-molecule inhibitor of MYC combined with PD-L1 blockade elicited a durable response in immune-cold TNBC with high MYC expression, suggesting a strategy to restore PD-L1 inhibitor sensitivity in MYC-overexpressing TNBC.


Subject(s)
Membrane Proteins/genetics , Proto-Oncogene Proteins c-myc/metabolism , Triple Negative Breast Neoplasms , Animals , B7-H1 Antigen , Cell Line, Tumor , Epigenetic Repression , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immune Evasion , Mice , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
10.
Clin J Am Soc Nephrol ; 16(5): 746-756, 2021 05 08.
Article in English | MEDLINE | ID: mdl-33858826

ABSTRACT

BACKGROUND AND OBJECTIVES: An accurate estimate of the loss of lifetime employment duration resulting from kidney failure can facilitate comprehensive evaluation of societal financial burdens. DESIGN, SETTING, PARTICIPANTS, & MEASUREMENTS: All patients undergoing incident dialysis in Taiwan during 2000-2017 were identified using the National Health Insurance Research Database. The corresponding age-, sex-, and calendar year-matched general population served as the referents. The survival functions and the employment states of the index cohort (patients on dialysis) and their referents for each age strata were first calculated, and then extrapolated until age 65 years, where the sum of the product of the survival function and the employment states was the lifetime employment duration. The difference in lifetime employment duration between the index and referent cohort was the loss of lifetime employment duration. Extrapolation of survival function and relative employment-to-population ratios were estimated by the restricted cubic spline models and the quadratic/linear models, respectively. RESULTS: A total of 83,358 patients with kidney failure were identified. Men had a higher rate of employment than women in each age strata. The expected loss of lifetime employment duration for men with kidney failure was 11.8, 7.6, 5.7, 3.8, 2.3, 1.0, and 0.2 years for those aged 25-34, 35-40, 41-45, 46-50, 51-55, 56-60, and 61-64 years, respectively; and the corresponding data for women was 10.5, 10.1, 7.9, 5.6, 3.3, 1.5, and 0.3 years, respectively. The values for loss of lifetime employment duration divided by loss of life expectancy were all >70% for women and >88% for men across the different age strata. The sensitivity analyses indicated that the results were robust. CONCLUSIONS: The loss of lifetime employment duration in patients undergoing dialysis mainly originates from loss of life expectancy.


Subject(s)
Employment/statistics & numerical data , Kidney Failure, Chronic/therapy , Life Expectancy , Renal Dialysis , Adult , Cohort Studies , Female , Humans , Male , Middle Aged , Taiwan , Time Factors
11.
Clin Cancer Res ; 27(15): 4379-4396, 2021 08 01.
Article in English | MEDLINE | ID: mdl-34011560

ABSTRACT

PURPOSE: FGFR1 overexpression has been associated with endocrine resistance in ER+ breast cancer. We found FGFR1 localized in the nucleus of breast cancer cells in primary tumors resistant to estrogen suppression. We investigated a role of nuclear FGFR1 on gene transcription and antiestrogen resistance. EXPERIMENTAL DESIGN: Tumors from patients treated with letrozole were subjected to Ki67 and FGFR1 IHC. MCF7 cells were transduced with FGFR1(SP-)(NLS) to promote nuclear FGFR1 overexpression. FGFR1 genomic activity in ER+/FGFR1-amplified breast cancer cells ± FOXA1 siRNA or ± the FGFR tyrosine kinase inhibitor (TKI) erdafitinib was examined by chromatin immunoprecipitation sequencing (ChIP-seq) and RNA sequencing (RNA-seq). The nuclear and chromatin-bound FGFR1 interactome was investigated by mass spectrometry (MS). RESULTS: High nuclear FGFR1 expression in ER+ primary tumors positively correlated with post-letrozole Ki67 values. Nuclear FGFR1 overexpression influenced gene transcription and promoted resistance to estrogen suppression and to fulvestrant in vivo. A gene expression signature induced by nuclear FGFR1 correlated with shorter survival in the METABRIC cohort of patients treated with antiestrogens. ChIP-Seq revealed FGFR1 occupancy at transcription start sites, overlapping with active transcription histone marks. MS analysis of the nuclear FGFR1 interactome identified phosphorylated RNA-Polymerase II and FOXA1, with FOXA1 RNAi impairing FGFR1 recruitment to chromatin. Treatment with erdafitinib did not impair nuclear FGFR1 translocation and genomic activity. CONCLUSIONS: These data suggest nuclear FGFR1 contributes to endocrine resistance by modulating gene transcription in ER+ breast cancer. Nuclear FGFR1 activity was unaffected by FGFR TKIs, thus supporting the development of treatment strategies to inhibit nuclear FGFR1 in ER+/FGFR1 overexpressing breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Estrogen Receptor Modulators/therapeutic use , Receptor, Fibroblast Growth Factor, Type 1/physiology , Transcription, Genetic/physiology , Breast Neoplasms/chemistry , Cell Nucleus , Female , Humans , Receptors, Estrogen/analysis , Tumor Cells, Cultured
12.
Cancer Cell ; 39(8): 1099-1114.e8, 2021 08 09.
Article in English | MEDLINE | ID: mdl-34171264

ABSTRACT

Activating mutations in HER2 (ERBB2) drive the growth of a subset of breast and other cancers and tend to co-occur with HER3 (ERBB3) missense mutations. The HER2 tyrosine kinase inhibitor neratinib has shown clinical activity against HER2-mutant tumors. To characterize the role of HER3 mutations in HER2-mutant tumors, we integrate computational structural modeling with biochemical and cell biological analyses. Computational modeling predicts that the frequent HER3E928G kinase domain mutation enhances the affinity of HER2/HER3 and reduces binding of HER2 to its inhibitor neratinib. Co-expression of mutant HER2/HER3 enhances HER2/HER3 co-immunoprecipitation and ligand-independent activation of HER2/HER3 and PI3K/AKT, resulting in enhanced growth, invasiveness, and resistance to HER2-targeted therapies, which can be reversed by combined treatment with PI3Kα inhibitors. Our results provide a mechanistic rationale for the evolutionary selection of co-occurring HER2/HER3 mutations and the recent clinical observations that HER3 mutations are associated with a poor response to neratinib in HER2-mutant cancers.


Subject(s)
Breast Neoplasms/genetics , Gain of Function Mutation , Quinolines/pharmacology , Receptor, ErbB-2/genetics , Receptor, ErbB-3/genetics , Aminopyridines/administration & dosage , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Humans , Mice, Nude , Molecular Docking Simulation , Molecular Dynamics Simulation , Morpholines/administration & dosage , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors/administration & dosage , Protein Multimerization , Quinolines/administration & dosage , Quinolines/chemistry , Quinolines/metabolism , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/chemistry , Receptor, ErbB-2/metabolism , Receptor, ErbB-3/chemistry , Receptor, ErbB-3/metabolism , Trastuzumab/pharmacology , Xenograft Model Antitumor Assays
13.
J Org Chem ; 75(15): 4921-8, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20578749

ABSTRACT

An efficient stereoselective synthesis of alpha-(2-->9)-tetrasialic acid was achieved using tri-O-chloroacetyl-derivatized sialyl donor and a triol sialyl acceptor. Both the acceptor and the donor were also protected with a cyclic 5-N-4-O-carbonyl protecting group. The donor is highly reactive and enabled alpha-selective sialylation with various primary, secondary, and tertiary acceptors under in situ activation conditions (NIS/TfOH, -78 degrees C, acetonitrile/dichloromethane). The trans-fused oxazolidinone ring and O-chloroacetyl protecting groups were easily removed under mild reaction conditions to provide the fully deprotected alpha(2-->9)-tetrasialic acid.

14.
Anal Bioanal Chem ; 398(2): 857-65, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20652556

ABSTRACT

The first liquid chromatography-tandem mass spectrometry method was developed and validated for the simultaneous quantification of p-aminohippuric acid and inulin, both typical biomarkers of kidney function. 5-(Hydroxymethyl)furfural, generated from inulin by acid and heat preparation, was used as an inulin substitute for the quantification. Acetaminophen was used as the internal standard. Solid-phase extraction was carried out with 5% methanol as the washing solution to optimize the retention of the analytes and to avoid obstruction of the orifice plate of the mass spectrometer caused by any unreacted inulin residue remaining from the sample preparation process. Chromatography separation was performed on a Symmetry C(18) column and a mobile phase composed of 2 mM ammonium formate and 0.1% formic acid in water (solvent A) and 2 mM ammonium formate and 0.1% formic acid in acetonitrile (solvent B) (30:70, v/v). Detection was performed with a triple-quadrupole tandem mass spectrometer using positive ion mode electrospray ionization in the multiple reaction monitoring mode. The selected transitions were m/z 195.2 → 120.2, 127.1 → 109.1, and 152.1 → 110.0 for p-aminohippuric acid, inulin [measured as 5-(hydroxymethyl)furfural], and acetaminophen, respectively. The linearity ranged from 10 to 140 µg/mL and from 100 to 1,400 µg/mL for p-aminohippurric acid and inulin (r > 0.99), respectively. The precisions and accuracies were all within 12 and 11% for the lower limit of quantification and quality control samples, respectively. This application was proven to be reliable and accurate and was successfully applied to a renal function study.


Subject(s)
Inulin/blood , Tandem Mass Spectrometry/methods , p-Aminohippuric Acid/blood , Animals , Chromatography, Liquid/methods , Kidney/metabolism , Kidney Function Tests , Male , Rats , Rats, Wistar , Sensitivity and Specificity , Solid Phase Extraction/methods
15.
Nat Commun ; 11(1): 5488, 2020 10 30.
Article in English | MEDLINE | ID: mdl-33127913

ABSTRACT

The 17q23 amplicon is associated with poor outcome in ER+ breast cancers, but the causal genes to endocrine resistance in this amplicon are unclear. Here, we interrogate transcriptome data from primary breast tumors and find that among genes in 17q23, PRR11 is a key gene associated with a poor response to therapeutic estrogen suppression. PRR11 promotes estrogen-independent proliferation and confers endocrine resistance in ER+ breast cancers. Mechanistically, the proline-rich motif-mediated interaction of PRR11 with the p85α regulatory subunit of PI3K suppresses p85 homodimerization, thus enhancing insulin-stimulated binding of p110-p85α heterodimers to IRS1 and activation of PI3K. PRR11-amplified breast cancer cells rely on PIK3CA and are highly sensitive to PI3K inhibitors, suggesting that PRR11 amplification confers PI3K dependence. Finally, genetic and pharmacological inhibition of PI3K suppresses PRR11-mediated, estrogen-independent growth. These data suggest ER+/PRR11-amplified breast cancers as a novel subgroup of tumors that may benefit from treatment with PI3K inhibitors and antiestrogens.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Estrogen Receptor Modulators/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proteins/genetics , Proteins/metabolism , Signal Transduction/drug effects , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Class I Phosphatidylinositol 3-Kinases/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm , Estrogen Antagonists/pharmacology , Estrogen Receptor Modulators/therapeutic use , Estrogens , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Insulin , Mice, Nude , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/therapeutic use , Xenograft Model Antitumor Assays
16.
J Chromatogr B Analyt Technol Biomed Life Sci ; 872(1-2): 58-62, 2008 Sep 01.
Article in English | MEDLINE | ID: mdl-18693079

ABSTRACT

A sensitive and selective liquid chromatography-tandem mass spectrometry (LC-MS/MS) method for the determination of triazolam and its metabolites, alpha-hydroxytriazolam (alpha-OHTRZ) and 4-hydroxytriazolam (4-OHTRZ), was developed and validated. Triazolam-D4 was used as the internal standard (IS). This analysis was carried out on a Thermo C(18) column and the mobile phase was composed of acetonitrile:H(2)O:formic acid (35:65:0.2, v/v/v). Detection was performed on a triple-quadrupole tandem mass spectrometer using positive ion mode electrospray ionization (ESI) and quantification was performed by multiple reaction monitoring (MRM) mode. The MS/MS ion transitions monitored were m/z 343.1-->308.3, 359.0-->308.3, 359.0-->111.2 and 347.0-->312.0 for triazolam, alpha-OHTRZ, 4-OHTRZ and triazolam-D4, respectively. LLOQ of the analytical method was 0.05 ng/mL for triazolam and 0.1 ng/mL for alpha-OHTRZ and 4-OHTRZ. The within- and between-run precisions were less than 15.26% and accuracy was -8.08% to 13.33%. The method proved to be accurate and specific, and was applied to the pharmacokinetic study of triazolam in healthy Chinese volunteers.


Subject(s)
Anti-Anxiety Agents/blood , Chromatography, High Pressure Liquid/methods , Tandem Mass Spectrometry/methods , Triazolam/blood , Anti-Anxiety Agents/pharmacokinetics , Humans , Reference Standards , Reproducibility of Results , Sensitivity and Specificity , Triazolam/pharmacokinetics
17.
Oncol Rep ; 39(6): 3041-3047, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29620216

ABSTRACT

Targeting cancer stem cells (CSCs) is a key strategy to prevent cancers from developing drug resistance and metastasis. Mitochondria have been reported to be a vulnerability of CSCs by multiple studies. Here, we report that doxycycline, functioning as an inhibitor of mitochondrial biogenesis, can effectively target breast cancer stem cells (BCSCs). Our results revealed that doxycycline significantly decreased the frequency of aldehyde dehydrogenase­positive (ALDH+) BCSCs as well as mammosphere formation efficiency in HER2+ and triple­negative breast cancer (TNBC) subtypes. Doxycycline also ameliorated paclitaxel­induced enrichment of ALDH+ BCSCs in TNBC. Mechanistically, we showed that doxycycline decreased the level of reactive oxygen species and their downstream p38 MAPK pathway. In agreement with the key role for p38 in maintaining BCSCs, a specific inhibitor targeting this MAPK pathway significantly decreased the number of ALDH+ cells. Doxycycline is a FDA­approved drug with minor and limited side­effects. Given doxycycline's low toxicity and strong effect on BCSC inhibition, we report that doxycycline should be safe to be used concomitantly with chemotherapy drugs to eradicate both CSCs and bulk tumor cells.


Subject(s)
Aldehyde Dehydrogenase/metabolism , Breast Neoplasms/metabolism , Doxycycline/pharmacology , Neoplastic Stem Cells/drug effects , Breast Neoplasms/drug therapy , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Neoplastic Stem Cells/metabolism , Reactive Oxygen Species/metabolism
18.
Cancer Lett ; 438: 165-173, 2018 12 01.
Article in English | MEDLINE | ID: mdl-30227220

ABSTRACT

Triple-negative breast cancer (TNBC) is the most difficult subtype of breast cancer to treat due to a paucity of effective targeted therapies. Many studies have reported that breast cancer stem cells (BCSCs) are enriched in TNBC and are responsible for chemoresistance and metastasis. In this study, we identify LRP8 as a novel positive regulator of BCSCs in TNBC. LRP8 is highly expressed in TNBC compared to other breast cancer subtypes and its genomic locus is amplified in 24% of TNBC tumors. Knockdown of LRP8 in TNBC cell lines inhibits Wnt/ß-catenin signaling, decreases BCSCs, and suppresses tumorigenic potential in xenograft models. LRP8 knockdown also induces a more differentiated, luminal-epithelial phenotype and thus sensitizes the TNBC cells to chemotherapy. Together, our study highlights LRP8 as a novel therapeutic target for TNBC as inhibition of LRP8 can attenuate Wnt/ß-catenin signaling to suppress BCSCs.


Subject(s)
Breast Neoplasms/genetics , LDL-Receptor Related Proteins/genetics , Neoplastic Stem Cells/metabolism , RNA Interference , Triple Negative Breast Neoplasms/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Cell Line, Tumor , Female , HEK293 Cells , Humans , Kaplan-Meier Estimate , LDL-Receptor Related Proteins/metabolism , Mice, Inbred NOD , Mice, SCID , RNAi Therapeutics/methods , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/therapy , Tumor Burden/genetics , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays/methods , beta Catenin/genetics
19.
Quintessence Int ; 48(3): 225-230, 2017.
Article in English | MEDLINE | ID: mdl-28168238

ABSTRACT

Numb chin syndrome (NCS) is a multifactorial neuropathic disorder associated with paresthesia to the chin, lip, and oral mucosa, particularly arising as a sequela to various dental-related procedures or infections in the mandible. Timely elucidation of the underlying etiology is of paramount importance as the presentation of NCS could serve as a harbinger of malignancy or metastatic disease. This report describes an unusual case of NCS developing synchronously with a vertical root fracture and odontogenic infection in a mandibular first molar. Clinicians should consider the inclusion of a vertical root fracture as plausible cofactor for the development of NCS.


Subject(s)
Chin , Dental Pulp Necrosis/diagnostic imaging , Dental Pulp Necrosis/surgery , Hypesthesia/etiology , Tooth Fractures/diagnostic imaging , Tooth Fractures/surgery , Tooth Root/injuries , Diagnosis, Differential , Humans , Magnetic Resonance Imaging , Middle Aged , Pain Measurement , Radiography, Panoramic , Syndrome , Tomography, X-Ray Computed , Tooth Extraction
20.
Carbohydr Res ; 341(11): 1948-53, 2006 Aug 14.
Article in English | MEDLINE | ID: mdl-16697995

ABSTRACT

Vanadyl triflate has been identified as a mild and efficient catalyst for the chemoselective O-isopropylidenation of functionalized carbohydrates with acetone and acetone equivalents. The current protocol is compatible with a diverse array of protecting groups and the products can be readily isolated by simple aqueous wash.


Subject(s)
Acetone/chemistry , Carbohydrates/chemistry , Mesylates/chemistry , Vanadates/chemistry , Catalysis , Glycosides/chemical synthesis , Glycosides/chemistry , Magnetic Resonance Spectroscopy , Molecular Structure
SELECTION OF CITATIONS
SEARCH DETAIL